1. Academic Validation
  2. RORα inhibits gastric cancer proliferation through attenuating G6PD and PFKFB3 induced glycolytic activity

RORα inhibits gastric cancer proliferation through attenuating G6PD and PFKFB3 induced glycolytic activity

  • Cancer Cell Int. 2024 Jan 6;24(1):12. doi: 10.1186/s12935-023-03201-4.
Xiaoshan Wang # 1 Junyi Zhang # 1 Yuwei Wu 1 Yuqing Zhang 2 Siyuan Zhang 1 Angqing Li 1 Jian Wang 1 Zhengguang Wang 3
Affiliations

Affiliations

  • 1 Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, 230032, Anhui, People's Republic of China.
  • 2 Department of Occupational Health and Environmental Hygiene, School of Public Health, Anhui Medical University, Hefei, Anhui, People's Republic of China.
  • 3 Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, 230032, Anhui, People's Republic of China. [email protected].
  • # Contributed equally.
Abstract

Background: Glycolysis is critical for harvesting abundant energy to maintain the tumor microenvironment in malignant tumors. Retinoic acid-related orphan receptor α (RORα) has been identified as a circadian gene. However, the association of glycolysis with RORα in regulating gastric Cancer (GC) proliferation remains poorly understood.

Methods: Bioinformatic analysis and retrospective study were utilized to explore the role of RORα in cell cycle and glycolysis in GC. The mechanisms were performed in vitro and in vivo including colony formation, Cell Counting Kit-8 (CCK-8), Epithelial- mesenchymal transition (EMT) and subcutaneous tumors of mice model assays. The key drives between RORα and glycolysis were verified through western blot and chip assays. Moreover, we constructed models of high proliferation and high glucose environments to verify a negative feedback and chemoresistance through a series of functional experiments in vitro and in vivo.

Results: RORα was found to be involved in the cell cycle and glycolysis through a gene set enrichment analysis (GSEA) algorithm. GC patients with low RORα expression were not only associated with high circulating tumor cells (CTC) and high vascular endothelial growth factor (VEGF) levels. However, it also presented a positive correlation with the standard uptake value (SUV) level. Moreover, the SUVmax levels showed a positive linear relation with CTC and VEGF levels. In addition, RORα expression levels were associated with glucose 6 phosphate dehydrogenase (G6PD) and phosphofructokinase-2/fructose-2,6-bisphosphatase (PFKFB3) expression levels, and GC patients with low RORα and high G6PD or low RORα and high PFKFB3 expression patterns had poorest disease-free survival (DFS). Functionally, RORα deletion promoted GC proliferation and drove glycolysis in vitro and in vivo. These phenomena were reversed by the RORα activator SR1078. Moreover, RORα deletion promoted GC proliferation through attenuating G6PD and PFKFB3 induced glycolytic activity in vitro and in vivo. Mechanistically, RORα was recruited to the G6PD and PFKFB3 promoters to modulate their transcription. Next, high proliferation and high glucose inhibited RORα expression, which indicated that negative feedback exists in GC. Moreover, RORα deletion improved fluorouracil chemoresistance through inhibition of glucose uptake.

Conclusion: RORα might be a novel biomarker and therapeutic target for GC through attenuating glycolysis.

Keywords

G6PD; Gastric cancer; Glycolysis; PFKFB3; Proliferation; RORα.

Figures
Products
  • Cat. No.
    Product Name
    Description
    Target
    Research Area
  • HY-19824
    99.56%, PFKFB3 Blocker